51
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
52
|
Delaby C, Hirtz C, Lehmann S. Overview of the blood biomarkers in Alzheimer's disease: Promises and challenges. Rev Neurol (Paris) 2023; 179:161-172. [PMID: 36371265 DOI: 10.1016/j.neurol.2022.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
The increasing number of people with advanced Alzheimer's disease (AD) represents a significant psychological and financial cost to the world population. Accurate detection of the earliest phase of preclinical AD is of major importance for the success of preventive and therapeutic strategies (Cullen et al., 2021). Advances in analytical techniques have been essential for the development of sensitive, specific and reliable diagnostic tests for AD biomarkers in biological fluids (cerebrospinal fluid and blood). Blood biomarkers hold promising potential for early and minimally invasive detection of AD, but also for differential diagnosis of dementia and for monitoring the course of the disease. The aim of this review is to provide an overview of current blood biomarkers of AD, from tau proteins and amyloid peptides to biomarkers of neuronal degeneration and inflammation, reactive and metabolic factors. We thus discuss the informative value of currently candidate blood biomarkers and their potential to be integrated into clinical practice for the management of AD in the near future.
Collapse
Affiliation(s)
- C Delaby
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France; Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Hirtz
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France
| | - S Lehmann
- LBPC-PPC, Université Montpellier, CHU Montpellier, INM Inserm, Montpellier, France.
| |
Collapse
|
53
|
Lazarev VF, Dutysheva EA, Kanunikov IE, Guzhova IV, Margulis BA. Protein Interactome of Amyloid-β as a Therapeutic Target. Pharmaceuticals (Basel) 2023; 16:312. [PMID: 37259455 PMCID: PMC9965366 DOI: 10.3390/ph16020312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 04/12/2024] Open
Abstract
The amyloid concept of Alzheimer's disease (AD) assumes the β-amyloid peptide (Aβ) as the main pathogenic factor, which injures neural and other brain cells, causing their malfunction and death. Although Aβ has been documented to exert its cytotoxic effect in a solitary manner, there is much evidence to claim that its toxicity can be modulated by other proteins. The list of such Aβ co-factors or interactors includes tau, APOE, transthyretin, and others. These molecules interact with the peptide and affect the ability of Aβ to form oligomers or aggregates, modulating its toxicity. Thus, the list of potential substances able to reduce the harmful effects of the peptide should include ones that can prevent the pathogenic interactions by specifically binding Aβ and/or its partners. In the present review, we discuss the data on Aβ-based complexes in AD pathogenesis and on the compounds directly targeting Aβ or the destructors of its complexes with other polypeptides.
Collapse
Affiliation(s)
- Vladimir F. Lazarev
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Elizaveta A. Dutysheva
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Igor E. Kanunikov
- Biological Faculty, St. Petersburg State University, 199034 Saint Petersburg, Russia
| | - Irina V. Guzhova
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Boris A. Margulis
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| |
Collapse
|
54
|
Moore JM, Bell EL, Hughes RO, Garfield AS. ABC transporters: human disease and pharmacotherapeutic potential. Trends Mol Med 2023; 29:152-172. [PMID: 36503994 DOI: 10.1016/j.molmed.2022.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are a 48-member superfamily of membrane proteins that actively transport a variety of biological substrates across lipid membranes. Their functional diversity defines an expansive involvement in myriad aspects of human biology. At least 21 ABC transporters underlie rare monogenic disorders, with even more implicated in the predisposition to and symptomology of common and complex diseases. Such broad (patho)physiological relevance places this class of proteins at the intersection of disease causation and therapeutic potential, underlining them as promising targets for drug discovery, as exemplified by the transformative CFTR (ABCC7) modulator therapies for cystic fibrosis. This review will explore the growing relevance of ABC transporters to human disease and their potential as small-molecule drug targets.
Collapse
|
55
|
de Frutos Lucas J, Sewell KR, García-Colomo A, Markovic S, Erickson KI, Brown BM. How does apolipoprotein E genotype influence the relationship between physical activity and Alzheimer's disease risk? A novel integrative model. Alzheimers Res Ther 2023; 15:22. [PMID: 36707869 PMCID: PMC9881295 DOI: 10.1186/s13195-023-01170-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/15/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Wide evidence suggests that physical activity (PA) confers protection against Alzheimer's disease (AD). On the other hand, the apolipoprotein E gene (APOE) ε4 allele represents the greatest genetic risk factor for developing AD. Extensive research has been conducted to determine whether frequent PA can mitigate the increased AD risk associated with APOE ε4. However, thus far, these attempts have produced inconclusive results. In this context, one possible explanation could be that the influence of the combined effect of PA and APOE ε4 carriage might be dependent on the specific outcome measure utilised. MAIN BODY In order to bridge these discrepancies, the aim of this theoretical article is to propose a novel model on the interactive effects of PA and APOE ε4 carriage on well-established mechanisms underlying AD. Available literature was searched to investigate how PA and APOE ε4 carriage, independently and in combination, may alter several molecular pathways involved in AD pathogenesis. The reviewed mechanisms include amyloid beta (Aβ) and tau deposition and clearance, neuronal resilience and neurogenesis, lipid function and cerebrovascular alterations, brain immune response and glucose metabolism. Finally, combining all this information, we have built an integrative model, which includes evidence-based and theoretical synergistic interactions across mechanisms. Moreover, we have identified key knowledge gaps in the literature, providing a list of testable hypotheses that future studies need to address. CONCLUSIONS We conclude that PA influences a wide array of molecular targets involved in AD neuropathology. A deeper understanding of where, when and, most importantly, how PA decreases AD risk even in the presence of the APOE ε4 allele will enable the creation of new protocols using exercise along pharmaceuticals in combined therapeutic approaches.
Collapse
Affiliation(s)
- Jaisalmer de Frutos Lucas
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain.
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
- Departamento de PsicologíaFacultad de Ciencias de la Vida y de la Naturaleza, Universidad Antonio de Nebrija, 28015, Madrid, Spain.
| | - Kelsey R Sewell
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Alejandra García-Colomo
- Experimental Psychology, Cognitive Processes and Logopedia Department, School of Psychology, Universidad Complutense de Madrid, 28223, Pozuelo de Alarcón, Spain
| | - Shaun Markovic
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071, Granada, Spain
- AdventHealth Research Institute, Orlando, FL, 32804, USA
| | - Belinda M Brown
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| |
Collapse
|
56
|
Aziz T, Nadeem AA, Sarwar A, Perveen I, Hussain N, Khan AA, Daudzai Z, Cui H, Lin L. Particle Nanoarchitectonics for Nanomedicine and Nanotherapeutic Drugs with Special Emphasis on Nasal Drugs and Aging. Biomedicines 2023; 11:354. [PMID: 36830891 PMCID: PMC9953552 DOI: 10.3390/biomedicines11020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/28/2023] Open
Abstract
Aging is a multifunctional physiological manifestation. The nasal cavity is considered a major site for easy and cost-effective drug and vaccine administration, due to high permeability, low enzymatic activity, and the presence of a high number of immunocompetent cells. This review article primarily focuses on aging genetics, physical parameters, and the use of nanoparticles as delivery systems of drugs and vaccines via the nasal cavity. Studies have identified various genes involved in centenarian and average-aged people. VEGF is a key mediator involved in angiogenesis. Different therapeutic approaches induce vascular function and angiogenesis. FOLR1 gene codes for folate receptor alpha protein that helps in regulating the transport of vitamin B folate, 5-methyltetrahydrofolate and folate analogs inside the cell. This gene also aids in slowing the aging process down by cellular regeneration and promotes healthy aging by reducing aging symptoms. It has been found through the literature that GATA 6, Yamanaka factors, and FOLR1 work in synchronization to induce healthy and delayed aging. The role and applications of genes including CBS, CISD, SIRT 1, and SIRT 6 play a significant role in aging.
Collapse
Affiliation(s)
- Tariq Aziz
- School of Food & Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Abad Ali Nadeem
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Abid Sarwar
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Ishrat Perveen
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Nageen Hussain
- Institute of Microbiology and Molecular Genetics, New Campus, University of the Punjab, Lahore 54590, Pakistan
| | - Ayaz Ali Khan
- Department of Biotechnology, University of Malakand, Chakdara 18800, Pakistan
| | - Zubaida Daudzai
- Department of Bioresources and Biotechnology, King Mongkut University of Technology, Bangkok 10140, Thailand
| | - Haiying Cui
- School of Food & Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Lin Lin
- School of Food & Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
57
|
Cross interactions between Apolipoprotein E and amyloid proteins in neurodegenerative diseases. Comput Struct Biotechnol J 2023; 21:1189-1204. [PMID: 36817952 PMCID: PMC9932299 DOI: 10.1016/j.csbj.2023.01.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Three common Apolipoprotein E isoforms, ApoE2, ApoE3, and ApoE4, are key regulators of lipid homeostasis, among other functions. Apolipoprotein E can interact with amyloid proteins. The isoforms differ by one or two residues at positions 112 and 158, and possess distinct structural conformations and functions, leading to isoform-specific roles in amyloid-based neurodegenerative diseases. Over 30 different amyloid proteins have been found to share similar characteristics of structure and toxicity, suggesting a common interactome. The molecular and genetic interactions of ApoE with amyloid proteins have been extensively studied in neurodegenerative diseases, but have not yet been well connected and clarified. Here we summarize essential features of the interactions between ApoE and different amyloid proteins, identify gaps in the understanding of the interactome and propose the general interaction mechanism between ApoE isoforms and amyloid proteins. Perhaps more importantly, this review outlines what we can learn from the interactome of ApoE and amyloid proteins; that is the need to see both ApoE and amyloid proteins as a basis to understand neurodegenerative diseases.
Collapse
|
58
|
Zhan N, Wang B, Martens N, Liu Y, Zhao S, Voortman G, van Rooij J, Leijten F, Vanmierlo T, Kuipers F, Jonker JW, Bloks VW, Lütjohann D, Palumbo M, Zimetti F, Adorni MP, Liu H, Mulder MT. Identification of Side Chain Oxidized Sterols as Novel Liver X Receptor Agonists with Therapeutic Potential in the Treatment of Cardiovascular and Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24021290. [PMID: 36674804 PMCID: PMC9863018 DOI: 10.3390/ijms24021290] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The nuclear receptors-liver X receptors (LXR α and β) are potential therapeutic targets in cardiovascular and neurodegenerative diseases because of their key role in the regulation of lipid homeostasis and inflammatory processes. Specific oxy(phyto)sterols differentially modulate the transcriptional activity of LXRs providing opportunities to develop compounds with improved therapeutic characteristics. We isolated oxyphytosterols from Sargassum fusiforme and synthesized sidechain oxidized sterol derivatives. Five 24-oxidized sterols demonstrated a high potency for LXRα/β activation in luciferase reporter assays and induction of LXR-target genes APOE, ABCA1 and ABCG1 involved in cellular cholesterol turnover in cultured cells: methyl 3β-hydroxychol-5-en-24-oate (S1), methyl (3β)-3-aldehydeoxychol-5-en-24-oate (S2), 24-ketocholesterol (S6), (3β,22E)-3-hydroxycholesta-5,22-dien-24-one (N10) and fucosterol-24,28 epoxide (N12). These compounds induced SREBF1 but not SREBP1c-mediated lipogenic genes such as SCD1, ACACA and FASN in HepG2 cells or astrocytoma cells. Moreover, S2 and S6 enhanced cholesterol efflux from HepG2 cells. All five oxysterols induced production of the endogenous LXR agonists 24(S)-hydroxycholesterol by upregulating the CYP46A1, encoding the enzyme converting cholesterol into 24(S)-hydroxycholesterol; S1 and S6 may also act via the upregulation of desmosterol production. Thus, we identified five novel LXR-activating 24-oxidized sterols with a potential for therapeutic applications in neurodegenerative and cardiovascular diseases.
Collapse
Affiliation(s)
- Na Zhan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Boyang Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Nikita Martens
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shangge Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Gardi Voortman
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Frank Leijten
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Johan W. Jonker
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Marcella Palumbo
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Correspondence: (H.L.); (M.T.M.)
| | - Monique T. Mulder
- Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
- Correspondence: (H.L.); (M.T.M.)
| |
Collapse
|
59
|
Young CB, Johns E, Kennedy G, Belloy ME, Insel PS, Greicius MD, Sperling RA, Johnson KA, Poston KL, Mormino EC. APOE effects on regional tau in preclinical Alzheimer's disease. Mol Neurodegener 2023; 18:1. [PMID: 36597122 PMCID: PMC9811772 DOI: 10.1186/s13024-022-00590-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND APOE variants are strongly associated with abnormal amyloid aggregation and additional direct effects of APOE on tau aggregation are reported in animal and human cell models. The degree to which these effects are present in humans when individuals are clinically unimpaired (CU) but have abnormal amyloid (Aβ+) remains unclear. METHODS We analyzed data from CU individuals in the Anti-Amyloid Treatment in Asymptomatic AD (A4) and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) studies. Amyloid PET data were available for 4486 participants (3163 Aβ-, 1323 Aβ+) and tau PET data were available for a subset of 447 participants (55 Aβ-, 392 Aβ+). Linear models examined APOE (number of e2 and e4 alleles) associations with global amyloid and regional tau burden in medial temporal lobe (entorhinal, amygdala) and early neocortical regions (inferior temporal, inferior parietal, precuneus). Consistency of APOE4 effects on regional tau were examined in 220 Aβ + CU and mild cognitive impairment (MCI) participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS APOE2 and APOE4 were associated with lower and higher amyloid positivity rates, respectively. Among Aβ+ CU, e2 and e4 were associated with reduced (-12 centiloids per allele) and greater (+15 centiloids per allele) continuous amyloid burden, respectively. APOE2 was associated with reduced regional tau in all regions (-0.05 to -0.09 SUVR per allele), whereas APOE4 was associated with greater regional tau (+0.02 to +0.07 SUVR per allele). APOE differences were confirmed by contrasting e3/e3 with e2/e3 and e3/e4. Mediation analyses among Aβ+ s showed that direct effects of e2 on regional tau were present in medial temporal lobe and early neocortical regions, beyond an indirect pathway mediated by continuous amyloid burden. For e4, direct effects on regional tau were only significant in medial temporal lobe. The magnitude of protective e2 effects on regional tau was consistent across brain regions, whereas detrimental e4 effects were greatest in medial temporal lobe. APOE4 patterns were confirmed in Aβ+ ADNI participants. CONCLUSIONS APOE influences early regional tau PET burden, above and beyond effects related to cross-sectional amyloid PET burden. Therapeutic strategies targeting underlying mechanisms related to APOE may modify tau accumulation among Aβ+ individuals.
Collapse
Affiliation(s)
- Christina B Young
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA.
| | - Emily Johns
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Gabriel Kennedy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Michael E Belloy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Philip S Insel
- University of California San Francisco, San Francisco, CA, USA
| | - Michael D Greicius
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Reisa A Sperling
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen L Poston
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Elizabeth C Mormino
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| |
Collapse
|
60
|
Walker JM, Gonzales MM, Goette W, Farrell K, White CL, Crary JF, Richardson TE. Cognitive and Neuropsychological Profiles in Alzheimer's Disease and Primary Age-Related Tauopathy and the Influence of Comorbid Neuropathologies. J Alzheimers Dis 2023; 92:1037-1049. [PMID: 36847012 PMCID: PMC11138480 DOI: 10.3233/jad-230022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
BACKGROUND Alzheimer's disease neuropathologic change (ADNC) is defined by the progression of both hyperphosphorylated-tau (p-tau) and amyloid-β (Aβ) and is the most common underlying cause of dementia worldwide. Primary age-related tauopathy (PART), an Aβ-negative tauopathy largely confined to the medial temporal lobe, is increasingly being recognized as an entity separate from ADNC with diverging clinical, genetic, neuroanatomic, and radiologic profiles. OBJECTIVE The specific clinical correlates of PART are largely unknown; we aimed to identify cognitive and neuropsychological differences between PART, ADNC, and subjects with no tauopathy (NT). METHODS We compared 2,884 subjects with autopsy-confirmed intermediate-high stage ADNC to 208 subjects with definite PART (Braak stage I-IV, Thal phase 0, CERAD NP score "absent") and 178 NT subjects from the National Alzheimer's Coordinating Center dataset. RESULTS PART subjects were older than either ADNC or NT patients. The ADNC cohort had more frequent neuropathological comorbidities as well as APOE ɛ4 alleles than the PART or NT cohort, and less frequent APOE ɛ2 alleles than either group. Clinically, ADNC patients performed significantly worse than NT or PART subjects across cognitive measures, but PART subjects had selective deficits in measures of processing speed, executive function, and visuospatial function, although additional cognitive measures were further impaired in the presence of neuropathologic comorbidities. In isolated cases of PART with Braak stage III-IV, there are additional deficits in measures of language. CONCLUSION Overall, these findings demonstrate underlying cognitive features specifically associated with PART, and reinforce the concept that PART is a distinct entity from ADNC.
Collapse
Affiliation(s)
- Jamie M. Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mitzi M. Gonzales
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - William Goette
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kurt Farrell
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles L. White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John F. Crary
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy E. Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
61
|
Iliyasu MO, Musa SA, Oladele SB, Iliya AI. Amyloid-beta aggregation implicates multiple pathways in Alzheimer's disease: Understanding the mechanisms. Front Neurosci 2023; 17:1081938. [PMID: 37113145 PMCID: PMC10128090 DOI: 10.3389/fnins.2023.1081938] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition characterized by tau pathology and accumulations of neurofibrillary tangles (NFTs) along with amyloid-beta (Aβ). It has been associated with neuronal damage, synaptic dysfunction, and cognitive deficits. The current review explained the molecular mechanisms behind the implications of Aβ aggregation in AD via multiple events. Beta (β) and gamma (γ) secretases hydrolyzed amyloid precursor protein (APP) to produce Aβ, which then clumps together to form Aβ fibrils. The fibrils increase oxidative stress, inflammatory cascade, and caspase activation to cause hyperphosphorylation of tau protein into neurofibrillary tangles (NFTs), which ultimately lead to neuronal damage. Acetylcholine (Ach) degradation is accelerated by upstream regulation of the acetylcholinesterase (AChE) enzyme, which leads to a deficiency in neurotransmitters and cognitive impairment. There are presently no efficient or disease-modifying medications for AD. It is necessary to advance AD research to suggest novel compounds for treatment and prevention. Prospectively, it might be reasonable to conduct clinical trials with unclean medicines that have a range of effects, including anti-amyloid and anti-tau, neurotransmitter modulation, anti-neuroinflammatory, neuroprotective, and cognitive enhancement.
Collapse
Affiliation(s)
- Musa O. Iliyasu
- Department of Anatomy, Kogi State University, Anyigba, Nigeria
- *Correspondence: Musa O. Iliyasu, ;
| | - Sunday A. Musa
- Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | - Sunday B. Oladele
- Department of Veterinary Pathology, Ahmadu Bello University, Zaria, Nigeria
| | | |
Collapse
|
62
|
Yuan Y, Yang B, He Y, Zhang W, E G. Genome-Wide Selection Signal Analysis of Australian Boer Goat by Insertion/Deletion Variants. RUSS J GENET+ 2022. [DOI: 10.1134/s1022795422120158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
63
|
Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G, Liu CC. ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol Neurodegener 2022; 17:72. [PMID: 36348357 PMCID: PMC9644639 DOI: 10.1186/s13024-022-00574-4] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia worldwide, and its prevalence is rapidly increasing due to extended lifespans. Among the increasing number of genetic risk factors identified, the apolipoprotein E (APOE) gene remains the strongest and most prevalent, impacting more than half of all AD cases. While the ε4 allele of the APOE gene significantly increases AD risk, the ε2 allele is protective relative to the common ε3 allele. These gene alleles encode three apoE protein isoforms that differ at two amino acid positions. The primary physiological function of apoE is to mediate lipid transport in the brain and periphery; however, additional functions of apoE in diverse biological functions have been recognized. Pathogenically, apoE seeds amyloid-β (Aβ) plaques in the brain with apoE4 driving earlier and more abundant amyloids. ApoE isoforms also have differential effects on multiple Aβ-related or Aβ-independent pathways. The complexity of apoE biology and pathobiology presents challenges to designing effective apoE-targeted therapeutic strategies. This review examines the key pathobiological pathways of apoE and related targeting strategies with a specific focus on the latest technological advances and tools.
Collapse
|
64
|
Wang L, Xiong Y, Fu B, Guo D, Zaky MY, Lin X, Wu H. MicroRNAs as immune regulators and biomarkers in tuberculosis. Front Immunol 2022; 13:1027472. [PMID: 36389769 PMCID: PMC9647078 DOI: 10.3389/fimmu.2022.1027472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 07/26/2023] Open
Abstract
Tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), is one of the most lethal infectious disease worldwide, and it greatly affects human health. Some diagnostic and therapeutic methods are available to effectively prevent and treat TB; however, only a few systematic studies have described the roles of microRNAs (miRNAs) in TB. Combining multiple clinical datasets and previous studies on Mtb and miRNAs, we state that pathogens can exploit interactions between miRNAs and other biomolecules to avoid host mechanisms of immune-mediated clearance and survive in host cells for a long time. During the interaction between Mtb and host cells, miRNA expression levels are altered, resulting in the changes in the miRNA-mediated regulation of host cell metabolism, inflammatory responses, apoptosis, and autophagy. In addition, differential miRNA expression can be used to distinguish healthy individuals, patients with TB, and patients with latent TB. This review summarizes the roles of miRNAs in immune regulation and their application as biomarkers in TB. These findings could provide new opportunities for the diagnosis and treatment of TB.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Yan Xiong
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Beibei Fu
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Dong Guo
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Mohamed Y. Zaky
- Department of Zoology, Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Xiaoyuan Lin
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| | - Haibo Wu
- Department of Biology, School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
65
|
Scarfò G, Piccarducci R, Daniele S, Franzoni F, Martini C. Exploring the Role of Lipid-Binding Proteins and Oxidative Stress in Neurodegenerative Disorders: A Focus on the Neuroprotective Effects of Nutraceutical Supplementation and Physical Exercise. Antioxidants (Basel) 2022; 11:2116. [PMID: 36358488 PMCID: PMC9686611 DOI: 10.3390/antiox11112116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 07/29/2023] Open
Abstract
The human brain is primarily composed of lipids, and their homeostasis is crucial to carry on normal neuronal functions. In order to provide an adequate amount of lipid transport in and out of the central nervous system, organisms need a set of proteins able to bind them. Therefore, alterations in the structure or function of lipid-binding proteins negatively affect brain homeostasis, as well as increase inflammation and oxidative stress with the consequent risk of neurodegeneration. In this regard, lifestyle changes seem to be protective against neurodegenerative processes. Nutraceutical supplementation with antioxidant molecules has proven to be useful in proving cognitive functions. Additionally, regular physical activity seems to protect neuronal vitality and increases antioxidant defenses. The aim of the present review was to investigate mechanisms that link lipid-binding protein dysfunction and oxidative stress to cognitive decline, also underlining the neuroprotective effects of diet and exercise.
Collapse
Affiliation(s)
- Giorgia Scarfò
- Department of Clinical and Experimental Medicine, Division of General Medicine, University of Pisa, 56126 Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, 56126 Pisa, Italy
| | | | - Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, Division of General Medicine, University of Pisa, 56126 Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, 56126 Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
66
|
Amyloid β, Lipid Metabolism, Basal Cholinergic System, and Therapeutics in Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms232012092. [PMID: 36292947 PMCID: PMC9603563 DOI: 10.3390/ijms232012092] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 12/05/2022] Open
Abstract
The presence of insoluble aggregates of amyloid β (Aβ) in the form of neuritic plaques (NPs) is one of the main features that define Alzheimer’s disease. Studies have suggested that the accumulation of these peptides in the brain significantly contributes to extensive neuronal loss. Furthermore, the content and distribution of cholesterol in the membrane have been shown to have an important effect on the production and subsequent accumulation of Aβ peptides in the plasma membrane, contributing to dysfunction and neuronal death. The monomeric forms of these membrane-bound peptides undergo several conformational changes, ranging from oligomeric forms to beta-sheet structures, each presenting different levels of toxicity. Aβ peptides can be internalized by particular receptors and trigger changes from Tau phosphorylation to alterations in cognitive function, through dysfunction of the cholinergic system. The goal of this review is to summarize the current knowledge on the role of lipids in Alzheimer’s disease and their relationship with the basal cholinergic system, as well as potential disease-modifying therapies.
Collapse
|
67
|
Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, Gouras GK, Deierborg T. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener 2022; 17:62. [PMID: 36153580 PMCID: PMC9509584 DOI: 10.1186/s13024-022-00566-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
ApoE is the major lipid and cholesterol carrier in the CNS. There are three major human polymorphisms, apoE2, apoE3, and apoE4, and the genetic expression of APOE4 is one of the most influential risk factors for the development of late-onset Alzheimer's disease (AD). Neuroinflammation has become the third hallmark of AD, together with Amyloid-β plaques and neurofibrillary tangles of hyperphosphorylated aggregated tau protein. This review aims to broadly and extensively describe the differential aspects concerning apoE. Starting from the evolution of apoE to how APOE's single-nucleotide polymorphisms affect its structure, function, and involvement during health and disease. This review reflects on how APOE's polymorphisms impact critical aspects of AD pathology, such as the neuroinflammatory response, particularly the effect of APOE on astrocytic and microglial function and microglial dynamics, synaptic function, amyloid-β load, tau pathology, autophagy, and cell-cell communication. We discuss influential factors affecting AD pathology combined with the APOE genotype, such as sex, age, diet, physical exercise, current therapies and clinical trials in the AD field. The impact of the APOE genotype in other neurodegenerative diseases characterized by overt inflammation, e.g., alpha- synucleinopathies and Parkinson's disease, traumatic brain injury, stroke, amyotrophic lateral sclerosis, and multiple sclerosis, is also addressed. Therefore, this review gathers the most relevant findings related to the APOE genotype up to date and its implications on AD and CNS pathologies to provide a deeper understanding of the knowledge in the APOE field.
Collapse
Affiliation(s)
- Rosalía Fernández-Calle
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Javier Frontiñán-Rubio
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Juan García-Revilla
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Martina Svensson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Isak Martinson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luís Venero
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Henrietta M. Nielsen
- Department of Biochemistry and Biophysics at, Stockholm University, Stockholm, Sweden
| | - Gunnar K. Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
68
|
Tomas-Roig J, Ramasamy S, Zbarsky D, Havemann-Reinecke U, Hoyer-Fender S. Psychosocial stress and cannabinoid drugs affect acetylation of α-tubulin (K40) and gene expression in the prefrontal cortex of adult mice. PLoS One 2022; 17:e0274352. [PMID: 36129937 PMCID: PMC9491557 DOI: 10.1371/journal.pone.0274352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
The dynamics of neuronal microtubules are essential for brain plasticity. Vesicular transport and synaptic transmission, additionally, requires acetylation of α-tubulin, and aberrant tubulin acetylation and neurobiological deficits are associated. Prolonged exposure to a stressor or consumption of drugs of abuse, like marihuana, lead to neurological changes and psychotic disorders. Here, we studied the effect of psychosocial stress and the administration of cannabinoid receptor type 1 drugs on α-tubulin acetylation in different brain regions of mice. We found significantly decreased tubulin acetylation in the prefrontal cortex in stressed mice. The impact of cannabinoid drugs on stress-induced microtubule disturbance was investigated by administration of the cannabinoid receptor agonist WIN55,212–2 and/or antagonist rimonabant. In both, control and stressed mice, the administration of WIN55,212–2 slightly increased the tubulin acetylation in the prefrontal cortex whereas administration of rimonabant acted antagonistically indicating a cannabinoid receptor type 1 mediated effect. The analysis of gene expression in the prefrontal cortex showed a consistent expression of ApoE attributable to either psychosocial stress or administration of the cannabinoid agonist. Additionally, ApoE expression inversely correlated with acetylated tubulin levels when comparing controls and stressed mice treated with WIN55,212–2 whereas rimonabant treatment showed the opposite.
Collapse
Affiliation(s)
- Jordi Tomas-Roig
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, Göttingen, Germany
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology–Developmental Biology, GZMB, Georg-August-University Göttingen, Göttingen, Germany
- * E-mail: (JTR); (SHF)
| | - Shyam Ramasamy
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology–Developmental Biology, GZMB, Georg-August-University Göttingen, Göttingen, Germany
| | - Diana Zbarsky
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology–Developmental Biology, GZMB, Georg-August-University Göttingen, Göttingen, Germany
| | - Ursula Havemann-Reinecke
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, Göttingen, Germany
| | - Sigrid Hoyer-Fender
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology–Developmental Biology, GZMB, Georg-August-University Göttingen, Göttingen, Germany
- * E-mail: (JTR); (SHF)
| |
Collapse
|
69
|
Almeida-Meza P, Richards M, Cadar D. Moderating Role of Cognitive Reserve Markers Between Childhood Cognition and Cognitive Aging: Evidence From the 1946 British Birth Cohort. Neurology 2022; 99:e1239-e1250. [PMID: 35922143 PMCID: PMC9576292 DOI: 10.1212/wnl.0000000000200928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES As the population ages, differences in cognitive abilities become more evident. We investigated key genetic and life course influences on cognitive state at age 69 years, building on previous work using the longitudinal Medical Research Council National Survey of Health and Development (the British 1946 birth cohort). METHODS Multivariable regressions investigated the association between 4 factors: (1) childhood cognition at age 8 years; (2) a Cognitive Reserve Index (CRI) composed of 3 markers: (i) educational attainment by age 26 years, (ii) engagement in leisure activities at age 43 years, and (iii) occupation up to age 53 years; (3) reading ability assessed by the National Adult Reading Test (NART) at age 53 years; and (4) APOE genotype in relation to cognitive state measured at age 69 years with Addenbrooke's Cognitive Examination, third edition (ACE-III). We then investigated the modifying role of the CRI, NART, and APOE in the association between childhood cognition and the ACE-III. RESULTS The analytical sample comprised 1,184 participants. Higher scores in childhood cognition, CRI, and NART were associated with higher scores in the ACE-III. We found that the CRI and NART modified the association between childhood cognition and the ACE-III: for 30 additional points in the CRI or 20 additional points in the NART, the simple slope of childhood cognition decreased by approximately 0.10 points (CRI = 70: marginal effects (MEs) 0.22, 95% CI 0.12-0.32, p < 0.001 vs CRI = 100: MEs 0.12, 95% CI 0.06-0.17, p < 0.001; NART = 15: MEs 0.22, 95% CI 0.09-0.35, p = 0.001, vs NART = 35: MEs 0.11, 95% CI 0.05-0.17, p < 0.001). The association between childhood cognition and the ACE-III was nonsignificant at high levels of the CRI or NART. Furthermore, the e4 allele of the APOE gene was associated with lower scores in the ACE-III (β = -0.71, 95% CI -1.36 to -0.06, p = 0.03) but did not modify the association between childhood cognition and cognitive state in later life. DISCUSSION The CRI and NART are independent measures of cognitive reserve because both modify the association between childhood cognition and cognitive state.
Collapse
Affiliation(s)
- Pamela Almeida-Meza
- From the Department of Behavioural Science and Health (P.A.-M., D.C.), University College London; MRC Unit for Lifelong Health and Ageing, University College London; and Centre for Dementia Studies (D.C.), Department of Neuroscience, Brighton and Sussex Medical School, UK.
| | - Marcus Richards
- From the Department of Behavioural Science and Health (P.A.-M., D.C.), University College London; MRC Unit for Lifelong Health and Ageing, University College London; and Centre for Dementia Studies (D.C.), Department of Neuroscience, Brighton and Sussex Medical School, UK
| | - Dorina Cadar
- From the Department of Behavioural Science and Health (P.A.-M., D.C.), University College London; MRC Unit for Lifelong Health and Ageing, University College London; and Centre for Dementia Studies (D.C.), Department of Neuroscience, Brighton and Sussex Medical School, UK
| |
Collapse
|
70
|
Liver X Receptor Regulation of Glial Cell Functions in the CNS. Biomedicines 2022; 10:biomedicines10092165. [PMID: 36140266 PMCID: PMC9496004 DOI: 10.3390/biomedicines10092165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
In this review, we discuss the role of liver X receptors (LXRs) in glial cells (microglia, oligodendrocytes and astrocytes) in the central nervous system (CNS). LXRs are oxysterol-activated nuclear receptors that, in adults, regulate genes involved in cholesterol homeostasis, the modulation of inflammatory responses and glutamate homeostasis. The study of LXR knockout mice has revealed that LXRβ plays a key role in maintaining the health of dopaminergic neurons in the substantia nigra, large motor neurons in the spinal cord and retinal ganglion cells in the eye. In the peripheral nervous system (PNS), LXRβ is responsible for the health of the spiral ganglion neurons (SGNs) in the cochlea. In addition, LXRs are essential for the homeostasis of the cerebrospinal fluid (CSF), and in LXRαβ−/− mice, the lateral ventricles are empty and lined with lipid-laden cells. As LXRαβ−/− mice age, lipid vacuoles accumulate in astrocytes surrounding blood vessels. By seven months of age, motor coordination becomes impaired, and there is a loss of motor neurons in the spinal cord of LXRβ−/− mice. During development, migration of neurons in the cortex and cerebellum is retarded in LXRβ−/− mice. Since LXRs are not expressed in dopaminergic or motor neurons in adult mice, the neuroprotective effects of LXRs appear to come from LXRs in glial cells where they are expressed. However, despite the numerous neurological deficits in LXR−/− rodents, multiple sclerosis has the clear distinction of being the only human neurodegenerative disease in which defective LXR signaling has been identified. In this review, we summarize the regulation and functions of LXRs in glial cells and analyze how targeting LXRs in glial cells might, in the future, be used to treat neurodegenerative diseases and, perhaps, disorders caused by aberrant neuronal migration during development.
Collapse
|
71
|
The Therapeutic Relevance of Urolithins, Intestinal Metabolites of Ellagitannin-Rich Food: A Systematic Review of In Vivo Studies. Nutrients 2022; 14:nu14173494. [PMID: 36079752 PMCID: PMC9460125 DOI: 10.3390/nu14173494] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 12/09/2022] Open
Abstract
The therapeutic effects of food rich in ellagitannins have been established to stem from its microbial metabolite, urolithin. Over the past decade, there has been a growing trend in urolithin research pertaining to its pharmacological properties. The purpose of this systematic review is to collate and synthesise all available data on urolithin’s therapeutic ability, to highlight its potential as a pharmaceutical agent, and prospective direction on future research. Methods: This systematic review was written based on the PRISMA guideline and was conducted across Ovid via Embase, Ovid MEDLINE, Cochrane Central Register for Controlled Trials, and Web of Science Core Collection. Results: A total of 41 animal studies were included in this systematic review based on the appropriate keyword. The included studies highlighted the neuroprotective, anti-metabolic disorder activity, nephroprotective, myocardial protective, anti-inflammatory, and musculoskeletal protection of urolithin A, B, and its synthetic analogue methylated urolithin A. The Sirt1, AMPK, and PI3K/AKT/mTOR signalling pathways were reported to be involved in the initiation of autophagy and mitochondrial biogenesis by urolithin A. Conclusions: This review methodically discusses the therapeutic prospects of urolithins and provides scientific justification for the potential development of urolithin A as a potent natural mitophagy inducer for anti-ageing purposes.
Collapse
|
72
|
Lipolysis-Stimulated Lipoprotein Receptor Acts as Sensor to Regulate ApoE Release in Astrocytes. Int J Mol Sci 2022; 23:ijms23158630. [PMID: 35955777 PMCID: PMC9368974 DOI: 10.3390/ijms23158630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022] Open
Abstract
Astroglia play an important role, providing de novo synthesized cholesterol to neurons in the form of ApoE-lipidated particles; disruption of this process can increase the risk of Alzheimer’s disease. We recently reported that glia-specific suppression of the lipolysis-stimulated lipoprotein receptor (LSR) gene leads to Alzheimer’s disease-like memory deficits. Since LSR is an Apo-E lipoprotein receptor, our objective of this study was to determine the effect of LSR expression modulation on cholesterol and ApoE output in mouse astrocytes expressing human ApoE3. qPCR analysis showed that siRNA-mediated lsr knockdown significantly increased expression of the genes involved in cholesterol synthesis, secretion, and metabolism. Analysis of media and lipoprotein fractions showed increased cholesterol and lipidated ApoE output in HDL-like particles. Further, lsr expression could be upregulated when astrocytes were incubated 5 days in media containing high levels (two-fold) of lipoprotein, or after 8 h treatment with 1 µM LXR agonist T0901317 in lipoprotein-deficient media. In both conditions of increased lsr expression, the ApoE output was repressed or unchanged despite increased abca1 mRNA levels and cholesterol production. We conclude that LSR acts as a sensor of lipoprotein content in the medium and repressor of ApoE release, while ABCA1 drives cholesterol efflux, thereby potentially affecting cholesterol load, ApoE lipidation, and limiting cholesterol trafficking towards the neuron.
Collapse
|
73
|
Zhang Y, Gao H, Zheng W, Xu H. Current understanding of the interactions between metal ions and Apolipoprotein E in Alzheimer's disease. Neurobiol Dis 2022; 172:105824. [PMID: 35878744 DOI: 10.1016/j.nbd.2022.105824] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/05/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease (AD), the most common type of dementia in the elderly, is a chronic and progressive neurodegenerative disorder with no effective disease-modifying treatments to date. Studies have shown that an imbalance in brain metal ions, such as zinc, copper, and iron, is closely related to the onset and progression of AD. Many efforts have been made to understand metal-related mechanisms and therapeutic strategies for AD. Emerging evidence suggests that interactions of brain metal ions and apolipoprotein E (ApoE), which is the strongest genetic risk factor for late-onset AD, may be one of the mechanisms for neurodegeneration. Here, we summarize the key points regarding how metal ions and ApoE contribute to the pathogenesis of AD. We further describe the interactions between metal ions and ApoE in the brain and propose that their interactions play an important role in neuropathological alterations and cognitive decline in AD.
Collapse
Affiliation(s)
- Yanhui Zhang
- Department of Tissue Engineering, China Medical University, Shenyang, China
| | - Huiling Gao
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Wei Zheng
- Department of Histology and Embryology, China Medical University, Shenyang, China
| | - He Xu
- Department of Anatomy, Histology and Embryology, School of Medicine, Shenzhen University, Shenzhen, China.
| |
Collapse
|
74
|
Abstract
The brain, as one of the most lipid-rich organs, heavily relies on lipid transport and distribution to maintain homeostasis and neuronal function. Lipid transport mediated by lipoprotein particles, which are complex structures composed of apolipoproteins and lipids, has been thoroughly characterized in the periphery. Although lipoproteins in the central nervous system (CNS) were reported over half a century ago, the identification of APOE4 as the strongest genetic risk factor for Alzheimer's disease has accelerated investigation of the biology and pathobiology of lipoproteins in the CNS. This review provides an overview of the different components of lipoprotein particles, in particular apolipoproteins, and their involvements in both physiological functions and pathological mechanisms in the CNS.
Collapse
Affiliation(s)
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| |
Collapse
|
75
|
Sacks F, Furtado J, Jensen M. Protein-based HDL subspecies: Rationale and association with cardiovascular disease, diabetes, stroke, and dementia. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159182. [DOI: 10.1016/j.bbalip.2022.159182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/09/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022]
|
76
|
Mai Y, Cao Z, Xu J, Yu Q, Yang S, Tang J, Zhao L, Fang W, Luo Y, Lei M, Mok VCT, Shi L, Liao W, Liu J. AD Resemblance Atrophy Index of Brain Magnetic Resonance Imaging in Predicting the Progression of Mild Cognitive Impairment Carrying Apolipoprotein E-ε4 Allele. Front Aging Neurosci 2022; 14:859492. [PMID: 35572149 PMCID: PMC9097868 DOI: 10.3389/fnagi.2022.859492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/23/2022] [Indexed: 01/03/2023] Open
Abstract
Background and Objective Early identification is important for timely Alzheimer’s disease (AD) treatment. Apolipoprotein E ε4 allele (APOE-ε4) is an important genetic risk factor for sporadic AD. The AD-Resemblance Atrophy Index (RAI)—a structural magnetic resonance imaging-derived composite index—was found to predict the risk of progression from mild cognitive impairment (MCI) to AD. Therefore, we investigated whether the AD-RAI can predict cognitive decline and progression to AD in patients with MCI carrying APOE ε4. Methods We included 733 participants with MCI from the Alzheimer’s Disease Neuroimaging Initiative Database (ADNI). Their APOE genotypes, cognitive performance, and levels of AD-RAI were assessed at baseline and follow-up. Linear regression models were used to test the correlations between the AD-RAI and baseline cognitive measures, and linear mixed models with random intercepts and slopes were applied to investigate whether AD-RAI and APOE-ε4 can predict the level of cognitive decline. Cox proportional risk regression models were used to test the association of AD-RAI and APOE status with the progression from MCI to AD. Results The baseline AD-RAI was higher in the MCI converted to AD group than in the MCI stable group (P < 0.001). The AD-RAI was significantly correlated with cognition, and had a synergistic effect with APOE-ε4 to predict the rate of cognitive decline. The AD-RAI predicted the risk and timing of MCI progression to AD. Based on the MCI population carrying APOE-ε4, the median time to progression from MCI to AD was 24 months if the AD-RAI > 0.5, while the median time to progression from MCI to AD was 96 months for patients with an AD-RAI ≤ 0.5. Conclusion The AD-RAI can predict the risk of progression to AD in people with MCI carrying APOE ε4, is strongly correlated with cognition, and can predict cognitive decline.
Collapse
Affiliation(s)
- Yingren Mai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyu Cao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qun Yu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaoqing Yang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingyi Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhao
- BrainNow Research Institute, Shenzhen, China
- BrainNow Medical Technology Limited, Shenzhen, China
| | - Wenli Fang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, China
- BrainNow Medical Technology Limited, Shenzhen, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Vincent C. T. Mok
- BrainNow Research Institute, Shenzhen, China
- BrainNow Medical Technology Limited, Shenzhen, China
- Division of Neurology, Department of Medicine and Therapeutics, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen, China
- BrainNow Medical Technology Limited, Shenzhen, China
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Wang Liao,
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Wang Liao,
| | | |
Collapse
|
77
|
Xin Y, Sheng J, Miao M, Wang L, Yang Z, Huang H. A review ofimaging genetics in Alzheimer's disease. J Clin Neurosci 2022; 100:155-163. [PMID: 35487021 DOI: 10.1016/j.jocn.2022.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/01/2022] [Accepted: 04/15/2022] [Indexed: 01/18/2023]
Abstract
Determining the association between genetic variation and phenotype is a key step to study the mechanism of Alzheimer's disease (AD), laying the foundation for studying drug therapies and biomarkers. AD is the most common type of dementia in the aged population. At present, three early-onset AD genes (APP, PSEN1, PSEN2) and one late-onset AD susceptibility gene apolipoprotein E (APOE) have been determined. However, the pathogenesis of AD remains unknown. Imaging genetics, an emerging interdisciplinary field, is able to reveal the complex mechanisms from the genetic level to human cognition and mental disorders via macroscopic intermediates. This paper reviews methods of establishing genotype-phenotype to explore correlations, including sparse canonical correlation analysis, sparse reduced rank regression, sparse partial least squares and so on. We found that most research work did poorly in supervised learning and exploring the nonlinear relationship between SNP-QT.
Collapse
Affiliation(s)
- Yu Xin
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| | - Jinhua Sheng
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China.
| | - Miao Miao
- Beijing Hospital, Beijing 100730, China; National Center of Gerontology, Beijing 100730, China; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Luyun Wang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China; Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
| | - Ze Yang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| | - He Huang
- College of Computer Science, Hangzhou Dianzi University, Hangzhou, Zhejiang 310018, China; Key Laboratory of Intelligent Image Analysis for Sensory and Cognitive Health, Ministry of Industry and Information Technology of China, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
78
|
Williams T, Ruiz AJ, Ruiz AM, Vo Q, Tsering W, Xu G, McFarland K, Giasson BI, Sullivan P, Borchelt DR, Chakrabarty P. Impact of APOE genotype on prion-type propagation of tauopathy. Acta Neuropathol Commun 2022; 10:57. [PMID: 35440098 PMCID: PMC9019935 DOI: 10.1186/s40478-022-01359-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Apolipoprotein (APOE) is a major risk factor of Alzheimer's disease (AD), with the E2, E3 and E4 isoforms differentially regulating the burden of AD-associated neuropathologies, such as amyloid β and tau. In AD, pathological tau is thought to spread along neuroanatomic connections following a prion-like mechanism. To provide insights into whether APOE isoforms differentially regulate the prion properties of tau and determine trans-synaptic transmission of tauopathy, we have generated human P301S mutant tau transgenic mice (PS19) that carry human APOE (APOE2, APOE3 or APOE4) or mouse Apoe allele. Mice received intrahippocamal injections of preformed aggregates of K18-tau at young ages, which were analyzed 5 months post-inoculation. Compared to the parental PS19 mice with mouse Apoe alleles, PS19 mice expressing human APOE alleles generally responded to K18-tau seeding with more intense AT8 immunoreactive phosphorylated tau athology. APOE3 homozygous mice accumulated higher levels of AT8-reactive ptau and microgliosis relative to APOE2 or APOE4 homozygotes (E3 > E4~2). PS19 mice that were heterozygous for APOE3 showed similar results, albeit to a lesser degree. In the timeframe of our investigation, we did not observe significant induction of argentophilic or MC1-reactive neurofibrillary tau tangle in PS19 mice homozygous for human APOE. To our knowledge, this is the first comprehensive study in rodent models that provides neuropathological insights into the dose-dependent effect of APOE isoforms on phosphorylated tau pathology induced by recombinant tau prions.
Collapse
|
79
|
Miron J, Picard C, Labonté A, Auld D, Poirier J. MSR1 and NEP Are Correlated with Alzheimer's Disease Amyloid Pathology and Apolipoprotein Alterations. J Alzheimers Dis 2022; 86:283-296. [PMID: 35034907 DOI: 10.3233/jad-215410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In mouse models of amyloidosis, macrophage receptor 1 (MSR1) and neprilysin (NEP) have been shown to interact to reduce amyloid burden in the brain. OBJECTIVE The purpose of this study is to analyze these two gene products in combination with apolipoproteins and Aβ1-42 in the cerebrospinal fluid (CSF) and plasma of individuals at different stages of Alzheimer's disease (AD), as well as in autopsied brain samples from ROSMAP (Religious Orders Study and Memory and Aging Project). METHODS CSF/plasma levels of MSR1 and NEP were measured using the sensitive primer extension assay technology. CSF Aβ1-42 was assessed with ELISA, while CSF ApoE and ApoJ were measured with the Luminex's multiplex technology. Brain MSR1, APOE, and CLU (APOJ) mRNA levels were measured with RNA-Seq and contrasted to amyloid plaques pathology using CERAD staging. RESULTS While plasma and CSF MSR1 levels are significantly correlated, this correlation was not observed for NEP. In addition to be highly correlated to one another, CSF levels of both MSR1 and NEP are strongly correlated with AD status and CSF Aβ1-42, ApoE, and ApoJ levels. In the cortical tissues of subjects from ROSMAP, MSR1 mRNA levels are correlated with CLU mRNA levels and the CERAD scores but not with APOE mRNA levels. CONCLUSION The discrepancies observed between CSF/plasma levels of MSR1 and NEP with CSF Aβ1-42 and ApoE concentrations can be explained by many factors, such as the disease stage or the involvement of the blood-brain barrier breakdown that leads to the infiltration of peripheral monocytes or macrophages.
Collapse
Affiliation(s)
- Justin Miron
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada.,McGill University, Montréal, QC, Canada
| | - Cynthia Picard
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Anne Labonté
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | | | - Judes Poirier
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada.,McGill University, Montréal, QC, Canada
| | | |
Collapse
|
80
|
Ondaro J, Hernandez-Eguiazu H, Garciandia-Arcelus M, Loera-Valencia R, Rodriguez-Gómez L, Jiménez-Zúñiga A, Goikolea J, Rodriguez-Rodriguez P, Ruiz-Martinez J, Moreno F, Lopez de Munain A, Holt IJ, Gil-Bea FJ, Gereñu G. Defects of Nutrient Signaling and Autophagy in Neurodegeneration. Front Cell Dev Biol 2022; 10:836196. [PMID: 35419363 PMCID: PMC8996160 DOI: 10.3389/fcell.2022.836196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/21/2022] [Indexed: 12/27/2022] Open
Abstract
Neurons are post-mitotic cells that allocate huge amounts of energy to the synthesis of new organelles and molecules, neurotransmission and to the maintenance of redox homeostasis. In neurons, autophagy is not only crucial to ensure organelle renewal but it is also essential to balance nutritional needs through the mobilization of internal energy stores. A delicate crosstalk between the pathways that sense nutritional status of the cell and the autophagic processes to recycle organelles and macronutrients is fundamental to guarantee the proper functioning of the neuron in times of energy scarcity. This review provides a detailed overview of the pathways and processes involved in the balance of cellular energy mediated by autophagy, which when defective, precipitate the neurodegenerative cascade of Parkinson's disease, frontotemporal dementia, amyotrophic lateral sclerosis or Alzheimer's disease.
Collapse
Affiliation(s)
- Jon Ondaro
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Haizea Hernandez-Eguiazu
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maddi Garciandia-Arcelus
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Raúl Loera-Valencia
- Department of Neurology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Laura Rodriguez-Gómez
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Andrés Jiménez-Zúñiga
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Julen Goikolea
- Department of Neurology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Patricia Rodriguez-Rodriguez
- Department of Neurology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Javier Ruiz-Martinez
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Donostia University Hospital, San Sebastian, Spain
| | - Fermín Moreno
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Donostia University Hospital, San Sebastian, Spain
| | - Adolfo Lopez de Munain
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Donostia University Hospital, San Sebastian, Spain
| | - Ian James Holt
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| | - Francisco Javier Gil-Bea
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Gorka Gereñu
- Department of Neuroscience, Biodonostia Health Research Institute (IIS Biodonostia), San Sebastian, Spain
- Center for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country (UPV-EHU), Leioa, Spain
| |
Collapse
|
81
|
Gharibyan AL, Wasana Jayaweera S, Lehmann M, Anan I, Olofsson A. Endogenous Human Proteins Interfering with Amyloid Formation. Biomolecules 2022; 12:biom12030446. [PMID: 35327638 PMCID: PMC8946693 DOI: 10.3390/biom12030446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 01/09/2023] Open
Abstract
Amyloid formation is a pathological process associated with a wide range of degenerative disorders, including Alzheimer’s disease, Parkinson’s disease, and diabetes mellitus type 2. During disease progression, abnormal accumulation and deposition of proteinaceous material are accompanied by tissue degradation, inflammation, and dysfunction. Agents that can interfere with the process of amyloid formation or target already formed amyloid assemblies are consequently of therapeutic interest. In this context, a few endogenous proteins have been associated with an anti-amyloidogenic activity. Here, we review the properties of transthyretin, apolipoprotein E, clusterin, and BRICHOS protein domain which all effectively interfere with amyloid in vitro, as well as displaying a clinical impact in humans or animal models. Their involvement in the amyloid formation process is discussed, which may aid and inspire new strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Anna L. Gharibyan
- Department of Clinical Microbiology, Umeå University, 901 87 Umeå, Sweden;
- Correspondence: (A.L.G.); (A.O.)
| | | | - Manuela Lehmann
- Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden; (M.L.); (I.A.)
| | - Intissar Anan
- Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden; (M.L.); (I.A.)
| | - Anders Olofsson
- Department of Clinical Microbiology, Umeå University, 901 87 Umeå, Sweden;
- Correspondence: (A.L.G.); (A.O.)
| |
Collapse
|
82
|
Lewandowski CT, Laham MS, Thatcher GR. Remembering your A, B, C's: Alzheimer's disease and ABCA1. Acta Pharm Sin B 2022; 12:995-1018. [PMID: 35530134 PMCID: PMC9072248 DOI: 10.1016/j.apsb.2022.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
The function of ATP binding cassette protein A1 (ABCA1) is central to cholesterol mobilization. Reduced ABCA1 expression or activity is implicated in Alzheimer's disease (AD) and other disorders. Therapeutic approaches to boost ABCA1 activity have yet to be translated successfully to the clinic. The risk factors for AD development and progression, including comorbid disorders such as type 2 diabetes and cardiovascular disease, highlight the intersection of cholesterol transport and inflammation. Upregulation of ABCA1 can positively impact APOE lipidation, insulin sensitivity, peripheral vascular and blood–brain barrier integrity, and anti-inflammatory signaling. Various strategies towards ABCA1-boosting compounds have been described, with a bias toward nuclear hormone receptor (NHR) agonists. These agonists display beneficial preclinical effects; however, important side effects have limited development. In particular, ligands that bind liver X receptor (LXR), the primary NHR that controls ABCA1 expression, have shown positive effects in AD mouse models; however, lipogenesis and unwanted increases in triglyceride production are often observed. The longstanding approach, focusing on LXRβ vs. LXRα selectivity, is over-simplistic and has failed. Novel approaches such as phenotypic screening may lead to small molecule NHR modulators that elevate ABCA1 function without inducing lipogenesis and are clinically translatable.
Collapse
|
83
|
Emerging role of HDL in brain cholesterol metabolism and neurodegenerative disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159123. [PMID: 35151900 DOI: 10.1016/j.bbalip.2022.159123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/07/2023]
Abstract
High-density lipoproteins (HDLs play a key role in cholesterol homeostasis maintenance in the central nervous system (CNS), by carrying newly synthesized cholesterol from astrocytes to neurons, to support their lipid-related physiological functions. As occurs for plasma HDLs, brain lipoproteins are assembled through the activity of membrane cholesterol transporters, undergo remodeling mediated by specific enzymes and transport proteins, and finally deliver cholesterol to neurons by a receptor-mediated internalization process. A growing number of evidences indicates a strong association between alterations of CNS cholesterol homeostasis and neurodegenerative disorders, in particular Alzheimer's disease (AD), and a possible role in this relationship may be played by defects in brain HDL metabolism. In the present review, we summarize and critically examine the current state of knowledge on major modifications of HDL and HDL-mediated brain cholesterol transport in AD, by taking into consideration the individual steps of this process. We also describe potential and encouraging HDL-based therapies that could represent new therapeutic strategies for AD treatment. Finally, we revise the main plasma and brain HDL modifications in other neurodegenerative disorders including Parkinson's disease (PD), Huntington's disease (HD), and frontotemporal dementia (FTD).
Collapse
|
84
|
Troutwine BR, Hamid L, Lysaker CR, Strope TA, Wilkins HM. Apolipoprotein E and Alzheimer's disease. Acta Pharm Sin B 2022; 12:496-510. [PMID: 35256931 PMCID: PMC8897057 DOI: 10.1016/j.apsb.2021.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic variation in apolipoprotein E (APOE) influences Alzheimer's disease (AD) risk. APOE ε4 alleles are the strongest genetic risk factor for late onset sporadic AD. The AD risk is dose dependent, as those carrying one APOE ε4 allele have a 2-3-fold increased risk, while those carrying two ε4 alleles have a 10-15-fold increased risk. Individuals carrying APOE ε2 alleles have lower AD risk and those carrying APOE ε3 alleles have neutral risk. APOE is a lipoprotein which functions in lipid transport, metabolism, and inflammatory modulation. Isoform specific effects of APOE within the brain include alterations to Aβ, tau, neuroinflammation, and metabolism. Here we review the association of APOE with AD, the APOE isoform specific effects within brain and periphery, and potential therapeutics.
Collapse
Affiliation(s)
- Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Laylan Hamid
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
85
|
Vitória JJM, Trigo D, da Cruz E Silva OAB. Revisiting APP secretases: an overview on the holistic effects of retinoic acid receptor stimulation in APP processing. Cell Mol Life Sci 2022; 79:101. [PMID: 35089425 PMCID: PMC11073327 DOI: 10.1007/s00018-021-04090-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide and is characterized by the accumulation of the β-amyloid peptide (Aβ) in the brain, along with profound alterations in phosphorylation-related events and regulatory pathways. The production of the neurotoxic Aβ peptide via amyloid precursor protein (APP) proteolysis is a crucial step in AD development. APP is highly expressed in the brain and is complexly metabolized by a series of sequential secretases, commonly denoted the α-, β-, and γ-cleavages. The toxicity of resulting fragments is a direct consequence of the first cleaving event. β-secretase (BACE1) induces amyloidogenic cleavages, while α-secretases (ADAM10 and ADAM17) result in less pathological peptides. Hence this first cleavage event is a prime therapeutic target for preventing or reverting initial biochemical events involved in AD. The subsequent cleavage by γ-secretase has a reduced impact on Aβ formation but affects the peptides' aggregating capacity. An array of therapeutic strategies are being explored, among them targeting Retinoic Acid (RA) signalling, which has long been associated with neuronal health. Additionally, several studies have described altered RA levels in AD patients, reinforcing RA Receptor (RAR) signalling as a promising therapeutic strategy. In this review we provide a holistic approach focussing on the effects of isoform-specific RAR modulation with respect to APP secretases and discuss its advantages and drawbacks in subcellular AD related events.
Collapse
Affiliation(s)
- José J M Vitória
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diogo Trigo
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Odete A B da Cruz E Silva
- Department of Medical Sciences, Neurosciences and Signalling Group, Institute of Biomedicine, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
86
|
Jain M, Singh MK, Shyam H, Mishra A, Kumar S, Kumar A, Kushwaha J. Role of JAK/STAT in the Neuroinflammation and its Association with Neurological Disorders. Ann Neurosci 2022; 28:191-200. [PMID: 35341232 PMCID: PMC8948319 DOI: 10.1177/09727531211070532] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Innate immunity is mediated by a variety of cell types, including microglia,
macrophages, and neutrophils, and serves as the immune system's first line of defense.
There are numerous pathways involved in innate immunity, including the interferon (IFN)
pathway, TRK pathway, mitogen-activated protein kinase (MAPK) pathway, Janus
kinase/signal transducer and activator of transcription (JAK/STAT) pathway, interleukin
(IL) pathways, chemokine pathways (CCR5), GSK signaling, and Fas signaling. Summary: JAK/STAT is one of these important signaling pathways and this review focused on
JAK/STAT signaling pathway only. The overactivation of microglia and astrocytes
influences JAK/STAT's role in neuroinflammatory disease by initiating innate immunity,
orchestrating adaptive immune mechanisms, and ultimately constraining inflammatory and
immunological responses. The JAK/STAT signaling pathway is one of the critical factors
that promotes neuroinflammation in neurodegenerative diseases. Key message: Given the importance of the JAK/STAT pathway in neurodegenerative disease, this review
discussed the feasibility of targeting the JAK/STAT pathway as a neuroprotective therapy
for neurodegenerative diseases in near future.
Collapse
Affiliation(s)
- Mayank Jain
- Department of Thoracic Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Mukul Kumar Singh
- Department of Urology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Hari Shyam
- Department of Thoracic Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Archana Mishra
- Department of Thoracic Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Shailendra Kumar
- Department of Thoracic Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Ambrish Kumar
- Department of Vascular Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Jitendra Kushwaha
- Department of General Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
87
|
Wang S, Sheng F, Zou L, Xiao J, Li P. Hyperoside attenuates non-alcoholic fatty liver disease in rats via cholesterol metabolism and bile acid metabolism. J Adv Res 2022; 34:109-122. [PMID: 35024184 PMCID: PMC8655136 DOI: 10.1016/j.jare.2021.06.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/05/2023] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) results from increased hepatic total cholesterol (TC) and total triglyceride (TG) accumulation. In our previous study, we found that rats treated with hyperoside became resistant to hepatic lipid accumulation. Objectives The present study aims to investigate the possible mechanisms responsible for the inhibitory effects of hyperoside on the lipid accumulation in the liver tissues of the NAFLD rats. Methods Label-free proteomics and metabolomics targeting at bile acid (BA) metabolism were applied to disclose the mechanisms for hyperoside reducing hepatic lipid accumulation among the NAFLD rats. Results In response to hyperoside treatment, several proteins related to the fatty acid degradation pathway, cholesterol metabolism pathway, and bile secretion pathway were altered, including ECI1, Acnat2, ApoE, and BSEP, etc. The expression of nuclear receptors (NRs), including farnesoid X receptor (FXR) and liver X receptor α (LXRα), were increased in hyperoside-treated rats' liver tissue, accompanied by decreased protein expression of catalyzing enzymes in the hepatic de novo lipogenesis and increased protein level of enzymes in the classical and alternative BA synthetic pathway. Liver conjugated BAs were less toxic and more hydrophilic than unconjugated BAs. The BA-targeted metabolomics suggest that hyperoside could decrease the levels of liver unconjugated BAs and increase the levels of liver conjugated BAs. Conclusions Taken together, the results suggest that hyperoside could improve the condition of NAFLD by regulating the cholesterol metabolism as well as BAs metabolism and excretion. These findings contribute to understanding the mechanisms by which hyperoside lowers the cholesterol and triglyceride in NAFLD rats.
Collapse
Key Words
- ACC, Acetyl-CoA carboxylase
- AMPK, AMP-activated protein kinase
- Apo, apolipoprotein
- BAs, bile acids
- BSH, bile salt hydrolase
- Bile acid metabolism
- CYP27A1, sterol 27-hydroxylase
- CYP7A1, cholesterol 7α-hydroxylase
- Cholesterol metabolism
- FGF15/19, fibroblast growth factor 15/19
- FXR, farnesoid X receptor
- Hyperoside
- LC-MS, the combination of high-performance liquid chromatography and mass spectrometry
- LXRα, liver X receptor α
- Label-free proteomics
- NAFLD
- NAFLD, non-alcoholic fatty liver disease
- PMSF, phenylmethylsulfonyl fluoride
- QC, quality control
- SDS, sodium dodecyl sulfate
- SHP, small heterodimer partner
- SREBP1, sterol regulatory element-binding protein 1
- SREBP2, sterol regulatory element-binding protein 2
- SREBPs, sterol regulatory element binding proteins
- TC, total cholesterol
- TG, triglyceride
- TGR5, Takeda G-protein-coupled receptor 5
- Targeted metabolomics
- VLDL, very low-density lipoprotein
- WB, Western blot
- pACC, phosphorylated ACC
Collapse
Affiliation(s)
- Songsong Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Feiya Sheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Liang Zou
- School of Medicine, Chengdu University, Chengdu 610106, China
| | - Jianbo Xiao
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China.,Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo, Vigo, Spain
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| |
Collapse
|
88
|
Parhizkar S, Holtzman DM. APOE mediated neuroinflammation and neurodegeneration in Alzheimer's disease. Semin Immunol 2022; 59:101594. [PMID: 35232622 PMCID: PMC9411266 DOI: 10.1016/j.smim.2022.101594] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/14/2022] [Indexed: 01/15/2023]
Abstract
Neuroinflammation is a central mechanism involved in neurodegeneration as observed in Alzheimer's disease (AD), the most prevalent form of neurodegenerative disease. Apolipoprotein E4 (APOE4), the strongest genetic risk factor for AD, directly influences disease onset and progression by interacting with the major pathological hallmarks of AD including amyloid-β plaques, neurofibrillary tau tangles, as well as neuroinflammation. Microglia and astrocytes, the two major immune cells in the brain, exist in an immune-vigilant state providing immunological defense as well as housekeeping functions that promote neuronal well-being. It is becoming increasingly evident that under disease conditions, these immune cells become progressively dysfunctional in regulating metabolic and immunoregulatory pathways, thereby promoting chronic inflammation-induced neurodegeneration. Here, we review and discuss how APOE and specifically APOE4 directly influences amyloid-β and tau pathology, and disrupts microglial as well as astroglial immunomodulating functions leading to chronic inflammation that contributes to neurodegeneration in AD.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease, Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease, Research Center, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
89
|
Dong L, Mao C, Liu C, Li J, Huang X, Wang J, Lei D, Chu S, Sha L, Xu Q, Peng B, Cui L, Gao J. Association Between Common Variants of APOE, ABCA7, A2M, BACE1, and Cerebrospinal Fluid Biomarkers in Alzheimer's Disease: Data from the PUMCH Dementia Cohort. J Alzheimers Dis 2021; 85:1511-1518. [PMID: 34958020 PMCID: PMC8925115 DOI: 10.3233/jad-215067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The previous studies have identified several genes in relation to Alzheimer’s disease (AD), such as ABCA7, CR1, etc. A few studies have explored the association between the common variants, mainly in the non-coding regions of these genes, and cerebrospinal fluid (CSF) biomarkers. Fewer studies target the variants in the coding regions. Objective: To illustrate the association between the common variants within or adjacent to the coding regions of AD susceptible genes and CSF biomarkers in AD patients. Methods: 75 sporadic probable AD patients were extracted from the dementia cohort of Peking Union Medical College Hospital. They all had history inquiry, physical examination, blood test, cognitive assessment, brain MRI, CSF testing of Aβ42, 181p-tau, and t-tau, and next-generation DNA sequencing. Sixty-nine common single nucleotide polymorphisms (SNPs) (minor allele frequency > 0.01) within or near the coding region of 13 AD susceptible genes were included in the analysis. Results: The rs7412-CC (APOE) genotype showed lower CSF Aβ42 level and higher p-tau/Aβ42 ratio than the rs7412-CT genotype. The rs3752246-C (ABCA7) allele correlated with lower CSF Aβ42 level. The alternate alleles of six ABCA7 SNPs were related to lower CSF p-tau, including rs3745842, rs3764648, rs3764652, rs4147930, rs4147934 and rs881768. The rs11609582-TT (A2M) genotype showed higher CSF p-tau than the rs11609582-TA genotype. The p-tau/Aβ42 ratio was higher in the rs490460-TT (BACE1) genotype relative to the rs490460-GT genotype. Conclusion: Some common variants within or near the coding regions of APOE, ABCA7, A2M, and BACE1 are associated with CSF Aβ42, p-tau. or p-tau/Aβ42.
Collapse
Affiliation(s)
- Liling Dong
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenhui Mao
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caiyan Liu
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Li
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinying Huang
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Lei
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shanshan Chu
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Longze Sha
- Institute of Basic Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qi Xu
- Institute of Basic Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bin Peng
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liying Cui
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Gao
- Neurology Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
90
|
Brain Expression, Physiological Regulation and Role in Motivation and Associative Learning of Peroxisome Proliferator-activated Receptor γ. Neuroscience 2021; 479:91-106. [PMID: 34762981 DOI: 10.1016/j.neuroscience.2021.10.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022]
Abstract
Like other members of the superfamily of nuclear receptors, the peroxisome proliferator-activated receptor γ (PPARγ), is a ligand-activated transcription factor known for its insulin-sensitizing actions in the periphery. Despite only sparse evidence for PPARγ in the CNS, many reports suggest direct PPARγ-mediated actions in the brain. This study aimed to (i) map PPARγ expression in rodent brain areas, involved in the regulation of cognitive, motivational, and emotional functions, (ii) examine the regulation of central PPARγ by physiological variables (age, sex, obesity); (iii) chemotypically identify PPARγ-expressing cells in the frontal cortex (FC) and hippocampus (HP); (iv) study whether activation of PPARγ by pioglitazone (Pio) in FC and HP cells can induce target gene expression; and (v) demonstrate the impact of activated PPARγ on learning behavior and motivation. Immunoreactive PPARγ was detectable in specific sub-nuclei/subfields of the FC, HP, nucleus accumbens, amygdala, hypothalamus, thalamus, and granular layers of the cerebellum. PPARγ protein levels were upregulated during aging and in high fat diet-induced obesity. PPARγ mRNA expression was upregulated in the amygdala of females (but not males) that were made obese. Neural precursor cells, mature neurons, and astrocytes in primary FC and HP cultures were shown to express PPARγ. Pioglitazone dose-dependently upregulated PPARγ target genes in manner that was specific to the origin (FC or HP) of the cultures. Lastly, administration of Pio impaired motivation and associative learning. Collectively, we provide evidence for the presence of regulatable PPARγ in the brain and demonstrate their participation the regulation of key behaviors.
Collapse
|
91
|
Katsuki H. Nuclear receptors of NR1 and NR4 subfamilies in the regulation of microglial functions and pathology. Pharmacol Res Perspect 2021; 9:e00766. [PMID: 34676987 PMCID: PMC8532137 DOI: 10.1002/prp2.766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
This review provides an overview of researches on the NR1 and NR4 nuclear receptors involved in the regulation of microglial functions. Nuclear receptors are attractive candidates for drug targets in the therapies of the central nervous system disorders, because the activation of these receptors is expected to regulate the functions and the phenotypes of microglia, by controlling the expression of specific gene subsets and also by regulating the cellular signaling mechanisms in a nongenomic manner. Several members of NR1 nuclear receptor subfamily have been examined for their ability to regulate microglial functions. For example, stimulation of vitamin D receptor inhibits the production of pro-inflammatory factors and increases the production of anti-inflammatory cytokines. Similar regulatory actions of nuclear receptor ligands on inflammation-related genes have also been reported for other NR1 members such as retinoic acid receptors, peroxisome proliferator-activated receptors (PPARs), and liver X receptors (LXRs). In addition, stimulation of PPARγ and LXRs may also result in increased phagocytic activities of microglia. Consistent with these actions, the agonists at nuclear receptors of NR1 subfamily are shown to produce therapeutic effects on animal models of various neurological disorders such as experimental allergic encephalomyelitis, Alzheimer's disease, Parkinson's disease, and ischemic/hemorrhagic stroke. On the other hand, increasing lines of evidence suggest that the stimulation of NR4 subfamily members of nuclear receptors such as Nur77 and Nurr1 also regulates microglial functions and alleviates neuropathological events in several disease models. Further advancement of these research fields may prove novel therapeutic opportunities.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico‐Pharmacological SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
92
|
Sharma NS, Karan A, Lee D, Yan Z, Xie J. Advances in Modeling Alzheimer's Disease In Vitro. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Anik Karan
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Donghee Lee
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Zheng Yan
- Department of Mechanical & Aerospace Engineering and Department of Biomedical Biological and Chemical Engineering University of Missouri Columbia MO 65211 USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
- Department of Mechanical and Materials Engineering College of Engineering University of Nebraska Lincoln Lincoln NE 68588 USA
| |
Collapse
|
93
|
Wakasugi N, Hanakawa T. It Is Time to Study Overlapping Molecular and Circuit Pathophysiologies in Alzheimer's and Lewy Body Disease Spectra. Front Syst Neurosci 2021; 15:777706. [PMID: 34867224 PMCID: PMC8637125 DOI: 10.3389/fnsys.2021.777706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia due to neurodegeneration and is characterized by extracellular senile plaques composed of amyloid β1 - 42 (Aβ) as well as intracellular neurofibrillary tangles consisting of phosphorylated tau (p-tau). Dementia with Lewy bodies constitutes a continuous spectrum with Parkinson's disease, collectively termed Lewy body disease (LBD). LBD is characterized by intracellular Lewy bodies containing α-synuclein (α-syn). The core clinical features of AD and LBD spectra are distinct, but the two spectra share common cognitive and behavioral symptoms. The accumulation of pathological proteins, which acquire pathogenicity through conformational changes, has long been investigated on a protein-by-protein basis. However, recent evidence suggests that interactions among these molecules may be critical to pathogenesis. For example, Aβ/tau promotes α-syn pathology, and α-syn modulates p-tau pathology. Furthermore, clinical evidence suggests that these interactions may explain the overlapping pathology between AD and LBD in molecular imaging and post-mortem studies. Additionally, a recent hypothesis points to a common mechanism of prion-like progression of these pathological proteins, via neural circuits, in both AD and LBD. This suggests a need for understanding connectomics and their alterations in AD and LBD from both pathological and functional perspectives. In AD, reduced connectivity in the default mode network is considered a hallmark of the disease. In LBD, previous studies have emphasized abnormalities in the basal ganglia and sensorimotor networks; however, these account for movement disorders only. Knowledge about network abnormalities common to AD and LBD is scarce because few previous neuroimaging studies investigated AD and LBD as a comprehensive cohort. In this paper, we review research on the distribution and interactions of pathological proteins in the brain in AD and LBD, after briefly summarizing their clinical and neuropsychological manifestations. We also describe the brain functional and connectivity changes following abnormal protein accumulation in AD and LBD. Finally, we argue for the necessity of neuroimaging studies that examine AD and LBD cases as a continuous spectrum especially from the proteinopathy and neurocircuitopathy viewpoints. The findings from such a unified AD and Parkinson's disease (PD) cohort study should provide a new comprehensive perspective and key data for guiding disease modification therapies targeting the pathological proteins in AD and LBD.
Collapse
Affiliation(s)
- Noritaka Wakasugi
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Hanakawa
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Integrated Neuroanatomy and Neuroimaging, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
94
|
Sharma N, Tan MA, An SSA. Phytosterols: Potential Metabolic Modulators in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms222212255. [PMID: 34830148 PMCID: PMC8618769 DOI: 10.3390/ijms222212255] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Phytosterols constitute a class of natural products that are an important component of diet and have vast applications in foods, cosmetics, and herbal medicines. With many and diverse isolated structures in nature, they exhibit a broad range of biological and pharmacological activities. Among over 200 types of phytosterols, stigmasterol and β-sitosterol were ubiquitous in many plant species, exhibiting important aspects of activities related to neurodegenerative diseases. Hence, this mini-review presented an overview of the reported studies on selected phytosterols related to neurodegenerative diseases. It covered the major phytosterols based on biosynthetic considerations, including other phytosterols with significant in vitro and in vivo biological activities.
Collapse
Affiliation(s)
- Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
| | - Mario A. Tan
- Research Center for the Natural and Applied Sciences, College of Science, University of Santo Tomas, Manila 1015, Philippines;
| | - Seong Soo A. An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
- Correspondence: ; Tel.: +82-31-750-8755
| |
Collapse
|
95
|
Ullah R, Park TJ, Huang X, Kim MO. Abnormal amyloid beta metabolism in systemic abnormalities and Alzheimer's pathology: Insights and therapeutic approaches from periphery. Ageing Res Rev 2021; 71:101451. [PMID: 34450351 DOI: 10.1016/j.arr.2021.101451] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated, multifactorial neurodegenerative disorder that is incurable. Despite recent success in treatments that partially improve symptomatic relief, they have failed in most clinical trials. Re-holding AD for accurate diagnosis and treatment is widely known as a challenging task. Lack of knowledge of basic molecular pathogenesis might be a possible reason for ineffective AD treatment. Historically, a majority of therapy-based studies have investigated the role of amyloid-β (Aβ peptide) in the central nervous system (CNS), whereas less is known about Aβ peptide in the periphery in AD. In this review, we provide a comprehensive summary of the current understanding of Aβ peptide metabolism (anabolism and catabolism) in the brain and periphery. We show that the abnormal metabolism of Aβ peptide is significantly linked with central-brain and peripheral abnormalities; the interaction between peripheral Aβ peptide metabolism and peripheral abnormalities affects central-brain Aβ peptide metabolism, suggesting the existence of significant communication between these two pathways of Aβ peptide metabolism. This close interaction between the central brain and periphery in abnormal Aβ peptide metabolism plays a key role in the development and progression of AD. In conclusion, we need to obtain a full understanding of the dynamic roles of Aβ peptide at the molecular level in both the brain and periphery in relation to the pathology of AD. This will not only provide new information regarding the complex disease pathology, but also offer potential new clues to improve therapeutic strategies and diagnostic biomarkers for the successful treatment of AD.
Collapse
|
96
|
Khodabakhsh P, Bazrgar M, Dargahi L, Mohagheghi F, Asgari Taei A, Parvardeh S, Ahmadiani A. Does Alzheimer's disease stem in the gastrointestinal system? Life Sci 2021; 287:120088. [PMID: 34715145 DOI: 10.1016/j.lfs.2021.120088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023]
Abstract
Over the last decades, our knowledge of the key pathogenic mechanisms of Alzheimer's disease (AD) has dramatically improved. Regarding the limitation of current therapeutic strategies for the treatment of multifactorial diseases, such as AD, to be translated into the clinic, there is a growing trend in research to identify risk factors associated with the onset and progression of AD. Here, we review the current literature with a focus on the relationship between gastrointestinal (GI)/liver diseases during the lifespan and the incidence of AD, and discuss the possible mechanisms underlying the link between the diseases. We also aim to review studies evaluating the possible link between the chronic use of the most common GI medications and the future risk of AD development.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Bazrgar
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohagheghi
- Institute of Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
97
|
Lee LY, Chou W, Chen WP, Wang MF, Chen YJ, Chen CC, Tung KC. Erinacine A-Enriched Hericium erinaceus Mycelium Delays Progression of Age-Related Cognitive Decline in Senescence Accelerated Mouse Prone 8 (SAMP8) Mice. Nutrients 2021; 13:nu13103659. [PMID: 34684662 PMCID: PMC8537498 DOI: 10.3390/nu13103659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 01/20/2023] Open
Abstract
There have been many reports on the neuroprotective effects of Hericium erinaceus mycelium, in which the most well-known active compounds found are diterpenoids, such as erinacine A. Previously, erinacine A-enriched Hericeum erinaceus mycelium (EAHEM) was shown to decrease amyloid plaque aggregation and improve cognitive disability in Alzheimer’s disease model APP/PS1 mice. However, its effects on brain aging have not yet been touched upon. Here, we used senescence accelerated mouse prone 8 (SAMP8) mice as a model to elucidate the mechanism by which EAHEM delays the aging of the brain. Three-month-old SAMP8 mice were divided into three EAHEM dosage groups, administered at 108, 215 and 431 mg/kg/BW/day, respectively. During the 12th week of EAHEM feeding, learning and memory of the mice were evaluated by single-trial passive avoidance and active avoidance test. After sacrifice, the amyloid plaques, induced nitric oxidase synthase (iNOS) activity, thiobarbituric acid-reactive substances (TBARS) and 8-OHdG levels were analyzed. We found that the lowest dose of 108 mg/kg/BW EAHEM was sufficient to significantly improve learning and memory in the passive and active avoidance tests. In all three EAHEM dose groups, iNOS, TBARS and 8-OHdG levels all decreased significantly and showed a dose-dependent response. The results indicate that EAHEM improved learning and memory and delayed degenerative aging in mice brains.
Collapse
Affiliation(s)
- Li-Ya Lee
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402204, Taiwan;
| | - Wayne Chou
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (W.C.); (W.-P.C.)
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (W.C.); (W.-P.C.)
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung 433303, Taiwan;
| | - Ying-Ju Chen
- College of Humanities & Social Sciences, Providence University, Taichung 433303, Taiwan;
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 106319, Taiwan;
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei 104336, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Kwong-Chung Tung
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402204, Taiwan;
- Correspondence:
| |
Collapse
|
98
|
Wołoszynowska-Fraser MU, Kouchmeshky A, McCaffery P. Vitamin A and Retinoic Acid in Cognition and Cognitive Disease. Annu Rev Nutr 2021; 40:247-272. [PMID: 32966186 DOI: 10.1146/annurev-nutr-122319-034227] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The history of vitamin A goes back over one hundred years, but our realization of its importance for the brain and cognition is much more recent. The brain is more efficient than other target tissues at converting vitamin A to retinoic acid (RA), which activates retinoic acid receptors (RARs). RARs regulate transcription, but their function in the cytoplasm to control nongenomic actions is also crucial. Controlled synthesis of RA is essential for regulating synaptic plasticity in regions of the brain involved in learning and memory, such as the hippocampus. Vitamin A deficiency results in a deterioration of these functions, and failure of RA signaling is perhaps associated with normal cognitive decline with age as well as with Alzheimer's disease. Further, several psychiatric and developmental disorders that disrupt cognition are also linked with vitamin A and point to their possible treatment with vitamin A or RA.
Collapse
Affiliation(s)
| | - Azita Kouchmeshky
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| |
Collapse
|
99
|
Chai AB, Lam HHJ, Kockx M, Gelissen IC. Apolipoprotein E isoform-dependent effects on the processing of Alzheimer's amyloid-β. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158980. [PMID: 34044125 DOI: 10.1016/j.bbalip.2021.158980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Since the identification of the apolipoprotein E (apoE) *ε4 allele as a major genetic risk factor for late-onset Alzheimer's disease, significant efforts have been aimed at elucidating how apoE4 expression confers greater brain amyloid-β (Aβ) burden, earlier disease onset and worse clinical outcomes compared to apoE2 and apoE3. ApoE primarily functions as a lipid carrier to regulate cholesterol metabolism in circulation as well as in the brain. However, it has also been suggested to interact with hydrophobic Aβ peptides to influence their processing in an isoform-dependent manner. Here, we review evidence from in vitro and in vivo studies extricating the effects of the three apoE isoforms, on different stages of the Aβ processing pathway including synthesis, aggregation, deposition, clearance and degradation. ApoE4 consistently correlates with impaired Aβ clearance, however data regarding Aβ synthesis and aggregation are conflicting and likely reflect inconsistencies in experimental approaches across studies. We further discuss the physical and chemical properties of apoE that may explain the inherent differences in activity between the isoforms. The lipidation status and lipid transport function of apoE are intrinsically linked with its ability to interact with Aβ. Traditionally, apoE-oriented therapeutic strategies for Alzheimer's disease have been proposed to non-specifically enhance or inhibit apoE activity. However, given the wide-ranging physiological functions of apoE in the brain and periphery, a more viable approach may be to specifically target and neutralise the pathological apoE4 isoform.
Collapse
Affiliation(s)
- Amanda B Chai
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Hin Hei Julian Lam
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord, NSW 2139, Australia
| | - Ingrid C Gelissen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
100
|
Peng HB, Bukuroshi P, Durk MR, Grootendorst P, Yan X, Pan SR, de Lannoy IAM, Pang KS. Impact of age, hypercholesterolemia, and the vitamin D receptor on brain endogenous β-amyloid peptide accumulation in mice. Biopharm Drug Dispos 2021; 42:372-388. [PMID: 34219248 DOI: 10.1002/bdd.2297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
Age, hypercholesterolemia, and vitamin D deficiency are risk factors that increase the brain accumulation of pathogenic β-amyloid peptides (40 and 42), precursors leading to Alzheimer's disease (AD) in humans. The relative changes accompanying aging, high cholesterol, and/or treatment of calcitriol, active vitamin D receptor (VDR) ligand, under normal physiology are unknown. We examined these relative changes in C57BL/6 mice of ages 2, 4-8, and more than 10 months old, which were fed a normal or high fat / high cholesterol diet and treated with calcitriol, active ligand of the vitamin D receptor (0 or 2.5 μg/kg ×4, intraperitoneally, every other day to elicit cholesterol lowering in liver). Aβ40 but not Aβ42 accumulation in brain and lower P-glycoprotein (P-gp) and neprilysin protein expressions for Aβ efflux and degradation, respectively, were found to be associated with aging. But there was no trend for BACE1 (β-secretase 1, a cholesterol-sensitive enzyme) toward Aβ synthesis with age. In response to calcitriol treatment, P-gp was elevated, mitigating partially the age-related changes. Although age-dependent decreasing trends in mRNA expression levels existed for Cyp46a1, the brain cholesterol processing enzyme, whose inhibition increases BACE1 and ApoE to facilitate microglia Aβ degradation, mRNA changes for other cholesterol transporters: Acat1 and Abca1, and brain cholesterol levels remained unchanged. There was no observable change in the mRNA expression of amyloid precursor protein (APP) and the influx (RAGE) and efflux (LRP1) transporters with respect to age, diet, or calcitriol treatment. Overall, aging poses as a risk factor contributing to Aβ accumulation in brain, and VDR-mediated P-gp activation partially alleviates the outcome.
Collapse
Affiliation(s)
- H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paola Bukuroshi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Matthew R Durk
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paul Grootendorst
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoyu Yan
- Department of Pharmacy, Chinese University of Hong Kong, Hong Kong, China
| | - Sophie R Pan
- InterVivo Solutions Inc., Mississauga, Ontario, Canada
| | | | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|